176 results on '"Todd C. McDevitt"'
Search Results
2. Hepatitis C virus infects and perturbs liver stem cells
- Author
-
Nathan L. Meyers, Tal Ashuach, Danielle E. Lyons, Mir M. Khalid, Camille R. Simoneau, Ann L. Erickson, Mehdi Bouhaddou, Thong T. Nguyen, G. Renuka Kumar, Taha Y. Taha, Vaishaali Natarajan, Jody L. Baron, Norma Neff, Fabio Zanini, Tokameh Mahmoudi, Stephen R. Quake, Nevan J. Krogan, Stewart Cooper, Todd C. McDevitt, Nir Yosef, and Melanie Ott
- Subjects
hepatitis c virus ,organoid ,liver disease ,stem cell ,single-cell RNA sequencing ,chronic infection ,Microbiology ,QR1-502 - Abstract
ABSTRACTHepatitis C virus (HCV) is the leading cause of death from liver disease. How HCV infection causes lasting liver damage and increases cancer risk remains unclear. Here, we identify bipotent liver stem cells as novel targets for HCV infection, and their erroneous differentiation as the potential cause of impaired liver regeneration and cancer development. We show 3D organoids generated from liver stem cells from actively HCV-infected individuals carry replicating virus and maintain low-grade infection over months. Organoids can be infected with a primary HCV isolate. Virus-inclusive single-cell RNA sequencing uncovered transcriptional reprogramming in HCV+ cells supporting hepatocytic differentiation, cancer stem cell development, and viral replication while stem cell proliferation and interferon signaling are disrupted. Our data add a new pathogenesis mechanism—infection of liver stem cells—to the biology of HCV infection that may explain progressive liver damage and enhanced cancer risk through an altered stem cell state.IMPORTANCEThe hepatitis C virus (HCV) causes liver disease, affecting millions. Even though we have effective antivirals that cure HCV, they cannot stop terminal liver disease. We used an adult stem cell-derived liver organoid system to understand how HCV infection leads to the progression of terminal liver disease. Here, we show that HCV maintains low-grade infections in liver organoids for the first time. HCV infection in liver organoids leads to transcriptional reprogramming causing cancer cell development and altered immune response. Our finding shows how HCV infection in liver organoids mimics HCV infection and patient pathogenesis. These results reveal that HCV infection in liver organoids contributes to liver disease progression.
- Published
- 2023
- Full Text
- View/download PDF
3. Diseased, differentiated and difficult: Strategies for improved engineering of in vitro neurological systems
- Author
-
Nicholas Elder, Faranak Fattahi, Todd C. McDevitt, and Lyandysha V. Zholudeva
- Subjects
stem cells ,cellular engineering ,directed neurons ,induced neurons ,micro RNA ,transcription factor complexes ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The rapidly growing field of cellular engineering is enabling scientists to more effectively create in vitro models of disease and develop specific cell types that can be used to repair damaged tissue. In particular, the engineering of neurons and other components of the nervous system is at the forefront of this field. The methods used to engineer neural cells can be largely divided into systems that undergo directed differentiation through exogenous stimulation (i.e., via small molecules, arguably following developmental pathways) and those that undergo induced differentiation via protein overexpression (i.e., genetically induced and activated; arguably bypassing developmental pathways). Here, we highlight the differences between directed differentiation and induced differentiation strategies, how they can complement one another to generate specific cell phenotypes, and impacts of each strategy on downstream applications. Continued research in this nascent field will lead to the development of improved models of neurological circuits and novel treatments for those living with neurological injury and disease.
- Published
- 2022
- Full Text
- View/download PDF
4. Modelling T-cell immunity against hepatitis C virus with liver organoids in a microfluidic coculture system
- Author
-
Vaishaali Natarajan, Camille R. Simoneau, Ann L. Erickson, Nathan L. Meyers, Jody L. Baron, Stewart Cooper, Todd C. McDevitt, and Melanie Ott
- Subjects
hepatitis C ,liver organoid ,CD8+ T cells ,microfluidics ,Biology (General) ,QH301-705.5 - Abstract
Hepatitis C virus (HCV) remains a global public health challenge with an estimated 71 million people chronically infected, with surges in new cases and no effective vaccine. New methods are needed to study the human immune response to HCV since in vivo animal models are limited and in vitro cancer cell models often show dysregulated immune and proliferative responses. Here, we developed a CD8+ T cell and adult stem cell liver organoid system using a microfluidic chip to coculture 3D human liver organoids embedded in extracellular matrix with HLA-matched primary human T cells in suspension. We then employed automated phase contrast and immunofluorescence imaging to monitor T cell invasion and morphological changes in the liver organoids. This microfluidic coculture system supports targeted killing of liver organoids when pulsed with a peptide specific for HCV non-structural protein 3 (NS3) (KLVALGINAV) in the presence of patient-derived CD8+ T cells specific for KLVALGINAV. This demonstrates the novel potential of the coculture system to molecularly study adaptive immune responses to HCV in an in vitro setting using primary human cells.
- Published
- 2022
- Full Text
- View/download PDF
5. Passive Clearing and 3D Lightsheet Imaging of the Intact and Injured Spinal Cord in Mice
- Author
-
Dylan A. McCreedy, Frank L. Jalufka, Madison E. Platt, Sun Won Min, Megan A. Kirchhoff, Anna L. Pritchard, Shelby K. Reid, Ronald Manlapaz, Eszter Mihaly, Jessica C. Butts, Nisha R. Iyer, Shelly E. Sakiyama-Elbert, Steven A. Crone, and Todd C. McDevitt
- Subjects
spinal cord injury ,lightsheet imaging ,PACT ,tissue clearing ,neural circuits ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The spinal cord contains a diverse array of sensory and motor circuits that are essential for normal function. Spinal cord injury (SCI) permanently disrupts neural circuits through initial mechanical damage, as well as a cascade of secondary injury events that further expand the spinal cord lesion, resulting in permanent paralysis. Tissue clearing and 3D imaging have recently emerged as promising techniques to improve our understanding of the complex neural circuitry of the spinal cord and the changes that result from damage due to SCI. However, the application of this technology for studying the intact and injured spinal cord remains limited. Here, we optimized the passive CLARITY technique (PACT) to obtain gentle and efficient clearing of the murine spinal cord without the need for specialized equipment. We demonstrate that PACT clearing enables 3D imaging of multiple fluorescent labels in the spinal cord to assess molecularly defined neuronal populations, acute inflammation, long-term tissue damage, and cell transplantation. Collectively, these procedures provide a framework for expanding the utility of tissue clearing to enhance the study of spinal cord neural circuits, as well as cellular- and tissue-level changes that occur following SCI.
- Published
- 2021
- Full Text
- View/download PDF
6. Allele-Specific Gene Editing Rescues Pathology in a Human Model of Charcot-Marie-Tooth Disease Type 2E
- Author
-
Carissa M. Feliciano, Kenneth Wu, Hannah L. Watry, Chiara B. E. Marley, Gokul N. Ramadoss, Hana Y. Ghanim, Angela Z. Liu, Lyandysha V. Zholudeva, Todd C. McDevitt, Mario A. Saporta, Bruce R. Conklin, and Luke M. Judge
- Subjects
induced pluripotent stem cells ,Charcot-Marie-Tooth ,neuropathy ,motor neurons ,dominant ,CRISPR-Cas9 ,Biology (General) ,QH301-705.5 - Abstract
Many neuromuscular disorders are caused by dominant missense mutations that lead to dominant-negative or gain-of-function pathology. This category of disease is challenging to address via drug treatment or gene augmentation therapy because these strategies may not eliminate the effects of the mutant protein or RNA. Thus, effective treatments are severely lacking for these dominant diseases, which often cause severe disability or death. The targeted inactivation of dominant disease alleles by gene editing is a promising approach with the potential to completely remove the cause of pathology with a single treatment. Here, we demonstrate that allele-specific CRISPR gene editing in a human model of axonal Charcot-Marie-Tooth (CMT) disease rescues pathology caused by a dominant missense mutation in the neurofilament light chain gene (NEFL, CMT type 2E). We utilized a rapid and efficient method for generating spinal motor neurons from human induced pluripotent stem cells (iPSCs) derived from a patient with CMT2E. Diseased motor neurons recapitulated known pathologic phenotypes at early time points of differentiation, including aberrant accumulation of neurofilament light chain protein in neuronal cell bodies. We selectively inactivated the disease NEFL allele in patient iPSCs using Cas9 enzymes to introduce a frameshift at the pathogenic N98S mutation. Motor neurons carrying this allele-specific frameshift demonstrated an amelioration of the disease phenotype comparable to that seen in an isogenic control with precise correction of the mutation. Our results validate allele-specific gene editing as a therapeutic approach for CMT2E and as a promising strategy to silence dominant mutations in any gene for which heterozygous loss-of-function is well tolerated. This highlights the potential for gene editing as a therapy for currently untreatable dominant neurologic diseases.
- Published
- 2021
- Full Text
- View/download PDF
7. Dynamic intercellular transport modulates the spatial patterning of differentiation during early neural commitment
- Author
-
Chad M. Glen, Todd C. McDevitt, and Melissa L. Kemp
- Subjects
Science - Abstract
How heterogeneities arise in stem cell populations remains unclear. Here, Glen et al. find that in ESC colonies cell cycle asynchronies modulate gap junctions, causing variation in intracellular signalling molecule diffusion between cells, and ultimately in spatial heterogeneity in differentiation.
- Published
- 2018
- Full Text
- View/download PDF
8. Perspective: The promise of multi-cellular engineered living systems
- Author
-
Roger D. Kamm, Rashid Bashir, Natasha Arora, Roy D. Dar, Martha U. Gillette, Linda G. Griffith, Melissa L. Kemp, Kathy Kinlaw, Michael Levin, Adam C. Martin, Todd C. McDevitt, Robert M. Nerem, Mark J. Powers, Taher A. Saif, James Sharpe, Shuichi Takayama, Shoji Takeuchi, Ron Weiss, Kaiming Ye, Hannah G. Yevick, and Muhammad H. Zaman
- Subjects
Biotechnology ,TP248.13-248.65 ,Medical technology ,R855-855.5 - Abstract
Recent technological breakthroughs in our ability to derive and differentiate induced pluripotent stem cells, organoid biology, organ-on-chip assays, and 3-D bioprinting have all contributed to a heightened interest in the design, assembly, and manufacture of living systems with a broad range of potential uses. This white paper summarizes the state of the emerging field of “multi-cellular engineered living systems,” which are composed of interacting cell populations. Recent accomplishments are described, focusing on current and potential applications, as well as barriers to future advances, and the outlook for longer term benefits and potential ethical issues that need to be considered.
- Published
- 2018
- Full Text
- View/download PDF
9. A rapid method for determining protein diffusion through hydrogels for regenerative medicine applications
- Author
-
Marian H. Hettiaratchi, Alex Schudel, Tel Rouse, Andrés J. García, Susan N. Thomas, Robert E. Guldberg, and Todd C. McDevitt
- Subjects
Biotechnology ,TP248.13-248.65 ,Medical technology ,R855-855.5 - Abstract
Hydrogels present versatile platforms for the encapsulation and delivery of proteins and cells for regenerative medicine applications. However, differences in hydrogel cross-linking density, polymer weight content, and affinity for proteins all contribute to diverse diffusion rates of proteins through hydrogel networks. Here, we describe a simple method to accurately measure protein diffusion through hydrogels, within a few hours and without the use of large amounts of protein. We tracked the diffusion of several proteins of varying molecular weights along the axial direction of capillary tubes filled with alginate, collagen, or poly(ethylene glycol) hydrogels. The rate of protein diffusion decreased with increasing molecular weight. A computational model of protein diffusion through capillary tubes was also created to predict and verify experimental protein diffusion coefficients. This in vitro capillary tube-based method of measuring protein diffusion represents a simple strategy to interrogate protein diffusion through natural and synthetic hydrogels and aid in the design of better biomaterial-based delivery vehicles that can effectively modulate protein release.
- Published
- 2018
- Full Text
- View/download PDF
10. Microscale Generation of Cardiospheres Promotes Robust Enrichment of Cardiomyocytes Derived from Human Pluripotent Stem Cells
- Author
-
Doan C. Nguyen, Tracy A. Hookway, Qingling Wu, Rajneesh Jha, Marcela K. Preininger, Xuemin Chen, Charles A. Easley, Paul Spearman, Shriprasad R. Deshpande, Kevin Maher, Mary B. Wagner, Todd C. McDevitt, and Chunhui Xu
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Cardiomyocytes derived from human pluripotent stem cells (hPSCs) are a promising cell source for regenerative medicine, disease modeling, and drug discovery, all of which require enriched cardiomyocytes, ideally ones with mature phenotypes. However, current methods are typically performed in 2D environments that produce immature cardiomyocytes within heterogeneous populations. Here, we generated 3D aggregates of cardiomyocytes (cardiospheres) from 2D differentiation cultures of hPSCs using microscale technology and rotary orbital suspension culture. Nearly 100% of the cardiospheres showed spontaneous contractility and synchronous intracellular calcium transients. Strikingly, from starting heterogeneous populations containing ∼10%–40% cardiomyocytes, the cell population within the generated cardiospheres featured ∼80%–100% cardiomyocytes, corresponding to an enrichment factor of up to 7-fold. Furthermore, cardiomyocytes from cardiospheres exhibited enhanced structural maturation in comparison with those from a parallel 2D culture. Thus, generation of cardiospheres represents a simple and robust method for enrichment of cardiomyocytes in microtissues that have the potential use in regenerative medicine as well as other applications.
- Published
- 2014
- Full Text
- View/download PDF
11. Author Correction: Dynamic intercellular transport modulates the spatial patterning of differentiation during early neural commitment
- Author
-
Chad M. Glen, Todd C. McDevitt, and Melissa L. Kemp
- Subjects
Science - Abstract
In the original version of this Article, an incorrect DOI number was provided in the Code Availability statement regarding the deposition of the computational model. The correct DOI is 10.5281/zenodo.1413539. This error has been corrected in both the PDF and HTML versions of the Article.
- Published
- 2018
- Full Text
- View/download PDF
12. Fabrication of and cell growth on 'silicon membranes' with high density TSVs for bio-sensing applications.
- Author
-
Muneeb Zia, Chaoqi Zhang 0001, Paragkumar Thadesar, Tracy Hookway, Taiyun Chi, Joe L. Gonzalez, Todd C. McDevitt, Hua Wang 0006, and Muhannad S. Bakir
- Published
- 2015
- Full Text
- View/download PDF
13. Cell culture and cell based sensor on CMOS.
- Author
-
Hua Wang 0006, Alborz Mahdavi, Jong Seok Park 0001, Taiyun Chi, Jessica Butts, Tracy A. Hookway, Todd C. McDevitt, David A. Tirrell, and Ali Hajimiri
- Published
- 2014
- Full Text
- View/download PDF
14. 11.7 A multimodality CMOS sensor array for cell-based assay and drug screening.
- Author
-
Jong Seok Park 0001, Taiyun Chi, Jessica Butts, Tracy Hookway, Todd C. McDevitt, and Hua Wang 0006
- Published
- 2015
- Full Text
- View/download PDF
15. A Multi-Modality CMOS Sensor Array for Cell-Based Assay and Drug Screening.
- Author
-
Taiyun Chi, Jong Seok Park 0001, Jessica C. Butts, Tracy A. Hookway, Amy Su, Chengjie Zhu, Mark P. Styczynski, Todd C. McDevitt, and Hua Wang 0006
- Published
- 2015
- Full Text
- View/download PDF
16. The NIH Somatic Cell Genome Editing program
- Author
-
Ross C. Wilson, Kevin D. Wells, W. Mark Saltzman, Philip J. Santangelo, Guohua Yi, Aravind Asokan, Shengdar Q. Tsai, Nenad Bursac, R. Holland Cheng, Shaoqin Gong, Gang Bao, Jennifer A. Doudna, Venkata S. Sabbisetti, Jarryd M. Campbell, Ryuji Morizane, Charles A. Gersbach, Mary E. Dickinson, Jon D. Hennebold, Kit S. Lam, Zheng-Yi Chen, John T. Hinson, Melinda R. Dwinell, Daniel G. Anderson, William R. Lagor, Qiaobing Xu, Melissa C. Skala, Jennifer A. Lewis, David J. Segal, Samantha Maragh, Guoping Feng, Stephen C. Ekker, Benjamin E. Deverman, Jonathan K. Watts, Alice F. Tarantal, Moriel H. Vandsburger, George A. Truskey, Ionita Ghiran, Marina E. Emborg, Jeff W.M. Bulte, Scot A. Wolfe, James E. Dahlman, Niren Murthy, Paul B. McCray, Erik J. Sontheimer, John C. Tilton, David T. Curiel, Benjamin S. Freedman, Guangping Gao, Mary Shimoyama, Kam W. Leong, Jiangbing Zhou, P. J. Brooks, Samira Kiani, Krystof S. Bankiewicz, Karl J. Clark, Jillian F. Banfield, Jon E. Levine, Krishanu Saha, Todd C. McDevitt, David R. Liu, Randall S. Prather, Daniel F. Carlson, Peter M. Glazer, Elliot L. Chaikof, Jason D. Heaney, Subhojit Roy, John A. Ronald, Stephen A. Murray, Cathleen M. Lutz, Anastasia Khvorova, Wen Xue, Sushmita Roy, Oleg Mirochnitchenko, Danith H. Ly, and David M. Gamm
- Subjects
Gene Editing ,Multidisciplinary ,Genome, Human ,Computer science ,Somatic cell ,Cells ,Targeted Gene Repair ,Genetic Therapy ,Computational biology ,Genome ,United States ,Targeted gene repair ,Human health ,National Institutes of Health (U.S.) ,Genome editing ,Research community ,Perspective ,Genetics research ,Animals ,Humans ,In patient ,Human genome ,Goals - Abstract
The move from reading to writing the human genome offers new opportunities to improve human health. The United States National Institutes of Health (NIH) Somatic Cell Genome Editing (SCGE) Consortium aims to accelerate the development of safer and more-effective methods to edit the genomes of disease-relevant somatic cells in patients, even in tissues that are difficult to reach. Here we discuss the consortium’s plans to develop and benchmark approaches to induce and measure genome modifications, and to define downstream functional consequences of genome editing within human cells. Central to this effort is a rigorous and innovative approach that requires validation of the technology through third-party testing in small and large animals. New genome editors, delivery technologies and methods for tracking edited cells in vivo, as well as newly developed animal models and human biological systems, will be assembled—along with validated datasets—into an SCGE Toolkit, which will be disseminated widely to the biomedical research community. We visualize this toolkit—and the knowledge generated by its applications—as a means to accelerate the clinical development of new therapies for a wide range of conditions., This Perspective discusses how the Somatic Cell Genome Editing Consortium aims to accelerate the implementation of safe and effective genome-editing therapies in the clinic.
- Published
- 2021
- Full Text
- View/download PDF
17. Spinal Interneurons as Gatekeepers to Neuroplasticity after Injury or Disease
- Author
-
Lyandysha V. Zholudeva, David S.K. Magnuson, Kajana Satkunendrarajah, Todd C. McDevitt, Michael A. Lane, Victoria E. Abraira, and Martyn Goulding
- Subjects
0301 basic medicine ,Traumatic spinal cord injury ,Interneuron ,Sensory system ,Disease ,Intact CNS ,03 medical and health sciences ,0302 clinical medicine ,Interneurons ,Neuroplasticity ,medicine ,Animals ,Humans ,GABA Agonists ,Spinal Cord Injuries ,Neurons ,Spinal interneuron ,Neuronal Plasticity ,business.industry ,musculoskeletal, neural, and ocular physiology ,General Neuroscience ,fungi ,Spinal cord ,030104 developmental biology ,medicine.anatomical_structure ,Spinal Cord ,nervous system ,Nervous System Diseases ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Spinal interneurons are important facilitators and modulators of motor, sensory, and autonomic functions in the intact CNS. This heterogeneous population of neurons is now widely appreciated to be a key component of plasticity and recovery. This review highlights our current understanding of spinal interneuron heterogeneity, their contribution to control and modulation of motor and sensory functions, and how this role might change after traumatic spinal cord injury. We also offer a perspective for how treatments can optimize the contribution of interneurons to functional improvement.
- Published
- 2021
- Full Text
- View/download PDF
18. Engineering human organoid development ex vivo—challenges and opportunities
- Author
-
Ana C. Silva, Todd C. McDevitt, and Oriane B. Matthys
- Subjects
0303 health sciences ,Computer science ,Cellular differentiation ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,02 engineering and technology ,Computational biology ,021001 nanoscience & nanotechnology ,Biomaterials ,03 medical and health sciences ,Organoid ,Stem cell ,0210 nano-technology ,030304 developmental biology - Abstract
The rapid progress of organoid technologies is attributable to the application of developmental biology principles, but organoid methods need further refinement provided by engineering approaches. However, we must first begin with common consensus on the critical features that distinguish organoids from simpler microtissues that lack the cellular complexity, structure, and function that organoids can achieve. Furthermore, current abilities to derive organoids from stem cells or multipotent progenitors favor certain germ lineages and tissue types more so than others, although the full reasons for this imbalance have yet to be determined. Technical challenges remain to identify the critical starting parameters for organoid reproducibility, systematically manipulate the proportions of differentiated cells from progenitors, and comprehensively characterize cell phenotypes spatially using advanced transcriptomic and 3D imaging methods. Advances in these regards will undoubtedly improve the robustness and predictability of existing organoids and permit the creation of organoids that have yet to be described.
- Published
- 2020
- Full Text
- View/download PDF
19. Self-Assembled Heterotypic Cardiac Spheroids from Human Pluripotent Stem Cells
- Author
-
Oriane B. Matthys and Todd C. McDevitt
- Published
- 2022
- Full Text
- View/download PDF
20. Hepatitis C Virus Infects and Perturbs Liver Stem Cells
- Author
-
Tal Ashuach, Danielle E Lyons, Jody L. Baron, Norma Neff, Mir M. Khalid, Camille R. Simoneau, Vaishaali Natarajan, Nir Yosef, Ann L Erickson, Thong T. Nguyen, Taha Y. Taha, Fabio Zanini, Stephen R. Quake, Melanie Ott, Nevan J. Krogan, Nathan L. Meyers, Mehdi Bouhaddou, Stewart Cooper, Todd C. McDevitt, and Tokameh Mahmoud
- Subjects
Hepatitis C virus ,Liver Stem Cell ,Biology ,medicine.disease ,medicine.disease_cause ,Virology ,Liver regeneration ,Liver disease ,Interferon ,Cancer stem cell ,medicine ,Stem cell ,Reprogramming ,medicine.drug - Abstract
SummaryHepatitis C virus (HCV) is the leading cause of death from liver disease. How HCV infection causes lasting liver damage and increases cancer risk beyond viral clearance remains unclear. We identify bipotent liver stem cells as novel targets for HCV infection, and their erroneous differentiation as the potential cause of impaired liver regeneration and cancer development. We show 3D organoids generated from liver stem cells from actively HCV-infected individuals carry replicating virus and maintain low-grade infection over months. Organoids can be infected with a primary HCV isolate. Virus-inclusive single-cell RNA-sequencing uncovered extensive transcriptional reprogramming in HCV+ cells supporting hepatocytic differentiation, cancer stem cell development and viral replication while stem cell proliferation and interferon signaling are disrupted. Our data adds a pathogenesis factor – infection of liver stem cells – to the biology of HCV infection that explains persistent liver damage and enhanced cancer risk through an altered stem cell state.
- Published
- 2021
- Full Text
- View/download PDF
21. Microfluidic perfusion modulates growth and motor neuron differentiation of stem cell aggregates
- Author
-
Amanda W. Schaefer, Emily Jackson-Holmes, Hang Lu, and Todd C. McDevitt
- Subjects
Cellular differentiation ,Microfluidics ,Cell ,Cell Culture Techniques ,Context (language use) ,01 natural sciences ,Biochemistry ,Article ,Analytical Chemistry ,03 medical and health sciences ,Electrochemistry ,medicine ,Environmental Chemistry ,Spectroscopy ,Microscale chemistry ,030304 developmental biology ,Motor Neurons ,0303 health sciences ,Chemistry ,010401 analytical chemistry ,Cell Differentiation ,Embryonic stem cell ,0104 chemical sciences ,Cell biology ,Perfusion ,medicine.anatomical_structure ,Cell culture ,Stem cell - Abstract
Microfluidic technologies provide many advantages for studying differentiation of three-dimensional (3D) stem cell aggregates, including the ability to control the culture microenvironment, isolate individual aggregates for longitudinal tracking, and perform imaging-based assays. However, applying microfluidics to studying mechanisms of stem cell differentiation requires an understanding of how microfluidic culture conditions impact cell phenotypes. Conventional cell culture techniques cannot directly be applied to the microscale, as microscale culture varies from macroscale culture in multiple aspects. Therefore, the objective of this work was to explore key parameters in microfluidic culture of 3D stem cell aggregates and to understand how these parameters influence stem cell behavior and differentiation. These studies were done in the context of differentiation of embryonic stem cells (ESCs) to motor neurons (MNs). We assessed how media exchange frequency modulates the biochemical microenvironment, including availability of exogenous factors (e.g., nutrients, small molecule additives) and cell-secreted molecules, and thereby impacts differentiation. The results of these studies provide guidance on how key characteristics of 3D cell cultures can be considered when designing microfluidic culture parameters. We demonstrate that discontinuous perfusion is effective at supporting stem cell aggregate growth. We find that there is a balance between the frequency of media exchange, which is needed to ensure that cells are not nutrient-limited, and the need to allow accumulation of cell-secreted factors to promote differentiation. Finally, we show how microfluidic device geometries can influence transport of biomolecules and potentially promote asymmetric spatial differentiation. These findings are instructive for future work in designing devices and experiments for culture of cell aggregates.
- Published
- 2020
- Full Text
- View/download PDF
22. Liver Organoid and T Cell Coculture Models Cytotoxic T Cell Responses against Hepatitis C Virus
- Author
-
Todd C. McDevitt, Nathan L. Meyers, Melanie Ott, Camille R. Simoneau, Stewart Cooper, Vaishaali Natarajan, Jody L. Baron, and Ann L Erickson
- Subjects
medicine.anatomical_structure ,Immune system ,T cell ,Hepatitis C virus ,Cancer cell ,Organoid ,Cancer research ,medicine ,Cytotoxic T cell ,Biology ,medicine.disease_cause ,CD8 ,Adult stem cell - Abstract
Hepatitis C virus (HCV) remains a global public health challenge with an estimated 71 million people chronically infected, with surges in new cases and no effective vaccine. New methods are needed to study the human immune response to HCV sincein vivoanimal models are limited andin vitrocancer cell models often show dysregulated immune and proliferative responses. Here we developed a CD8+T cell and adult stem cell liver organoid system using a microfluidic chip to coculture 3D human liver organoids embedded in extracellular matrix with HLA-matched primary human T cells in suspension. We then employed automated phase contrast and immunofluorescence imaging to monitor T cell invasion and morphological changes in the liver organoids. This microfluidic coculture system supports targeted killing of liver organoids when pulsed with a peptide specific for HCV nonstructural protein 3 (NS3) (KLVALGINAV) in the presence of patient-derived CD8+T cells specific for KLVALGINAV. This demonstrates the novel potential of the coculture system to molecularly study adaptive immune responses to HCV in anin vitrosetting using primary human cells.
- Published
- 2021
- Full Text
- View/download PDF
23. Developmental lineage of human pluripotent stem cell‐derived cardiac fibroblasts affects their functional phenotype
- Author
-
Charles M. Kerr, Timothy J. Kamp, Brenda M. Ogle, Martha E. Floy, Alexandra B. Steinberg, Sophie E. Givens, Ana C. Silva, Jianhua Zhang, Ying Mei, Oriane B. Matthys, Sean P. Palecek, Taylor D. Mateyka, and Todd C. McDevitt
- Subjects
Pluripotent Stem Cells ,Transcription, Genetic ,Biology ,Biochemistry ,Article ,Extracellular matrix ,Paracrine signalling ,Genetics ,medicine ,Humans ,Cell Lineage ,Myocytes, Cardiac ,Secretion ,Myofibroblasts ,Fibroblast ,Induced pluripotent stem cell ,Molecular Biology ,Cells, Cultured ,Myocardium ,Cell Differentiation ,Phenotype ,Extracellular Matrix ,Cell biology ,medicine.anatomical_structure ,sense organs ,Myofibroblast ,Function (biology) ,Biotechnology - Abstract
Cardiac fibroblasts (CFBs) support heart function by secreting extracellular matrix (ECM) and paracrine factors, respond to stress associated with injury and disease, and therefore are an increasingly important therapeutic target. We describe how developmental lineage of human pluripotent stem cell-derived CFBs, epicardial (EpiC-FB), and second heart field (SHF-FB) impacts transcriptional and functional properties. Both EpiC-FBs and SHF-FBs exhibited CFB transcriptional programs and improved calcium handling in human pluripotent stem cell-derived cardiac tissues. We identified differences including in composition of ECM synthesized, secretion of growth and differentiation factors, and myofibroblast activation potential, with EpiC-FBs exhibiting higher stress-induced activation potential akin to myofibroblasts and SHF-FBs demonstrating higher calcification and mineralization potential. These phenotypic differences suggest that EpiC-FBs have utility in modeling fibrotic diseases while SHF-FBs are a promising source of cells for regenerative therapies. This work directly contrasts regional and developmental specificity of CFBs and informs CFB in vitro model selection.
- Published
- 2021
- Full Text
- View/download PDF
24. Axial elongation of caudalized human organoids mimics aspects of neural tube development
- Author
-
Eliza A Gaylord, Ashley R.G. Libby, Todd C. McDevitt, Jessica C Butts, Martina Z Krakora, Nicholas H Elder, David A Joy, Emily A Bulger, and Frederico N. Mendoza-Camacho
- Subjects
Pluripotent Stem Cells ,Neural Tube ,Neurogenesis ,Organogenesis ,Embryonic Development ,Hindbrain ,Biology ,Mesoderm ,SOX2 ,medicine ,Organoid ,Humans ,Hox gene ,Molecular Biology ,Wnt Signaling Pathway ,Body Patterning ,Neural tube ,Wnt signaling pathway ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Stem Cells and Regeneration ,Cell biology ,Neuroepithelial cell ,Organoids ,medicine.anatomical_structure ,PAX6 ,Developmental Biology - Abstract
Axial elongation of the neural tube is crucial during mammalian embryogenesis for anterior-posterior body axis establishment and subsequent spinal cord development, but these processes cannot be interrogated directly in humans as they occur post-implantation. Here, we report an organoid model of neural tube extension derived from human pluripotent stem cell (hPSC) aggregates that have been caudalized with Wnt agonism, enabling them to recapitulate aspects of the morphological and temporal gene expression patterns of neural tube development. Elongating organoids consist largely of neuroepithelial compartments and contain TBXT+SOX2+ neuro-mesodermal progenitors in addition to PAX6+NES+ neural progenitors. A critical threshold of Wnt agonism stimulated singular axial extensions while maintaining multiple cell lineages, such that organoids displayed regionalized anterior-to-posterior HOX gene expression with hindbrain (HOXB1) regions spatially distinct from brachial (HOXC6) and thoracic (HOXB9) regions. CRISPR interference-mediated silencing of TBXT, a Wnt pathway target, increased neuroepithelial compartmentalization, abrogated HOX expression and disrupted uniaxial elongation. Together, these results demonstrate the potent capacity of caudalized hPSC organoids to undergo axial elongation in a manner that can be used to dissect the cellular organization and patterning decisions that dictate early human nervous system development.
- Published
- 2021
25. Live demonstration: A multi-modality CMOS sensor array for cell-based assay and drug screening.
- Author
-
Jong Seok Park 0001, Taiyun Chi, Amy Su, Chengjie Zhu, Jessica Butts, Tracy Hookway, Todd C. McDevitt, Mark P. Styczynski, and Hua Wang 0006
- Published
- 2015
- Full Text
- View/download PDF
26. Passive clearing and 3D lightsheet imaging of the intact and injured spinal cord in mice
- Author
-
Nisha Iyer, Ron Manlapaz, Jessica C Butts, Shelly E. Sakiyama-Elbert, Frank L. Jalufka, Dylan A. McCreedy, Steven A. Crone, Sun Won Min, Eszter Mihaly, Todd C. McDevitt, Megan A. Kirchhoff, and Madison E. Platt
- Subjects
Tissue clearing ,business.industry ,Sensory system ,medicine.disease ,Spinal cord ,Cell transplantation ,medicine.anatomical_structure ,medicine ,Paralysis ,Biological neural network ,Spinal cord lesion ,medicine.symptom ,business ,Neuroscience ,Spinal cord injury - Abstract
The spinal cord contains a diverse array of sensory and motor circuits that are essential for normal function. Spinal cord injury (SCI) permanently disrupts neural circuits through initial mechanical damage, as well as a cascade of secondary injury events that further expand the spinal cord lesion, resulting in permanent paralysis. Tissue clearing and 3D imaging have recently emerged as promising techniques to improve our understanding of the complex neural circuitry of the spinal cord and the changes that result from damage due to SCI. However, the application of this technology for studying the intact and injured spinal cord remains limited. Here we optimized the passive CLARITY technique (PACT) to obtain gentle and efficient clearing of the murine spinal cord without the need for specialized equipment. We demonstrate that PACT clearing enables 3D imaging of multiple fluorescent labels in the spinal cord to assess molecularly defined neuronal populations, acute inflammation, long-term tissue damage, and cell transplantation. Collectively, these procedures provide a framework for expanding the utility of tissue clearing to enhance the study of spinal cord neural circuits, as well as cellular- and tissue-level changes that occur following SCI.
- Published
- 2021
- Full Text
- View/download PDF
27. Spatial Pattern Dynamics of 3D Stem Cell Loss of Pluripotency via Rules-Based Computational Modeling.
- Author
-
Douglas E. White, Melissa A. Kinney, Todd C. McDevitt, and Melissa L. Kemp
- Published
- 2013
- Full Text
- View/download PDF
28. Engineering the Spatiotemporal Mosaic Self-Patterning of Pluripotent Stem Cells
- Author
-
Ashley R.G. Libby, Todd C. McDevitt, and David A Joy
- Subjects
0303 health sciences ,Gene knockdown ,CRISPR interference ,Morphogenesis ,Biology ,Cell biology ,03 medical and health sciences ,Multicellular organism ,0302 clinical medicine ,Live cell imaging ,Cell tracking ,Induced pluripotent stem cell ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Pluripotent stem cells (PSCs) possess the ability to self-organize into complex tissue-like structures; however, the genetic mechanisms and multicellular dynamics that direct such patterning are difficult to control. Here, we pair live imaging with controlled induction of gene knockdown by CRISPR interference (CRISPRi) to generate changes within subpopulations of human PSCs, allowing for control over organization and analysis of emergent behaviors. Specifically, we use forced aggregation of mixtures of cells with and without an inducible CRISPRi system to knockdown molecular regulators of tissue symmetry. We then track the resulting multicellular organization through fluorescence live imaging concurrent with the induction of knockdown. Overall, this technique allows for controlled initiation of symmetry breaking by CRISPRi to produce changes in cellular behavior that can be tracked over time within high-density pluripotent stem cell colonies.
- Published
- 2020
- Full Text
- View/download PDF
29. Engineering the Spatiotemporal Mosaic Self-Patterning of Pluripotent Stem Cells
- Author
-
Ashley R G, Libby, David A, Joy, and Todd C, McDevitt
- Subjects
Gene Editing ,Pluripotent Stem Cells ,Microscopy, Video ,Time Factors ,CRISPR-Associated Proteins ,Gene Expression Regulation, Developmental ,Time-Lapse Imaging ,Microscopy, Fluorescence ,Clustered Regularly Interspaced Short Palindromic Repeats ,CRISPR-Cas Systems ,Cells, Cultured ,Body Patterning ,RNA, Guide, Kinetoplastida ,Signal Transduction - Abstract
Pluripotent stem cells (PSCs) possess the ability to self-organize into complex tissue-like structures; however, the genetic mechanisms and multicellular dynamics that direct such patterning are difficult to control. Here, we pair live imaging with controlled induction of gene knockdown by CRISPR interference (CRISPRi) to generate changes within subpopulations of human PSCs, allowing for control over organization and analysis of emergent behaviors. Specifically, we use forced aggregation of mixtures of cells with and without an inducible CRISPRi system to knockdown molecular regulators of tissue symmetry. We then track the resulting multicellular organization through fluorescence live imaging concurrent with the induction of knockdown. Overall, this technique allows for controlled initiation of symmetry breaking by CRISPRi to produce changes in cellular behavior that can be tracked over time within high-density pluripotent stem cell colonies.
- Published
- 2020
30. SARS-CoV-2 infection of human iPSC-derived cardiac cells reflects cytopathic features in hearts of patients with COVID-19
- Author
-
David A Joy, Bruce M. McManus, Ken Nakamura, Melanie Ott, Camille R. Simoneau, Ana C. Silva, Parinaz Fozouni, Will R. Flanigan, Pei-Yi Chen, Bruce R. Conklin, Paul J. Hanson, Gokul N. Ramadoss, Huihui Li, Serah Kang, Juan A. Perez-Bermejo, Sarah J. Rockwood, Jeffrey D. Whitman, and Todd C. McDevitt
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Induced Pluripotent Stem Cells ,Autopsy ,030204 cardiovascular system & hematology ,Asymptomatic ,Article ,Pathogenesis ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Myocyte ,Humans ,Myocytes, Cardiac ,Fragmentation (cell biology) ,Induced pluripotent stem cell ,Cells, Cultured ,business.industry ,SARS-CoV-2 ,Myocardium ,Structural gene ,COVID-19 ,Heart ,General Medicine ,030104 developmental biology ,medicine.symptom ,business - Abstract
Although coronavirus disease 2019 (COVID-19) causes cardiac dysfunction in up to 25% of patients, its pathogenesis remains unclear. Exposure of human induced pluripotent stem cell (iPSC)-derived heart cells to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) revealed productive infection and robust transcriptomic and morphological signatures of damage, particularly in cardiomyocytes. Transcriptomic disruption of structural genes corroborates adverse morphologic features, which included a distinct pattern of myofibrillar fragmentation and nuclear disruption. Human autopsy specimens from patients with COVID-19 reflected similar alterations, particularly sarcomeric fragmentation. These notable cytopathic features in cardiomyocytes provide insights into SARS-CoV-2-induced cardiac damage, offer a platform for discovery of potential therapeutics, and raise concerns about the long-term consequences of COVID-19 in asymptomatic and severe cases.
- Published
- 2020
31. Silencing of E-cadherin in induced human pluripotent stem cells promotes extraembryonic fates accompanying multilineage differentiation
- Author
-
Ashley R.G. Libby, Todd C. McDevitt, David A Joy, Bruce R. Conklin, Frederico N. Mendoza-Camacho, Ivana Vasic, and Martina Z Krakora
- Subjects
medicine.anatomical_structure ,Cadherin ,Cell ,Embryogenesis ,Morphogenesis ,medicine ,Germ layer ,Biology ,Cell fate determination ,Induced pluripotent stem cell ,Embryonic stem cell ,Cell biology - Abstract
Summary/AbstractIn embryonic development, symmetry breaking events and the mechanical milieus in which they occur coordinate the specification of separate cell lineages. Here, we use 3D aggregates of human pluripotent stem cells (hPSCs) encapsulated in alginate microbeads to model the early blastocyst prior to zona pellucida hatching. We demonstrate that 3D confinement combined with modulation of cell-cell adhesions is sufficient to drive differentiation and collective migration reminiscent of the pre-implantation embryo. Knockdown of the cell adhesion protein CDH1 in encapsulated hPSC aggregates resulted in protrusion morphologies and emergence of extra-embryonic lineages, whereas unencapsulated CDH1(-) aggregates displayed organized radial delamination and mesendoderm specification bias. Transcriptomic similarities between single-cell RNA-sequencing data of early human embryos and encapsulated CDH1(-) aggregates establishes this in vitro system as a competent surrogate for studying early embryonic fate decisions and highlights the relationship between cell-cell adhesions and the mechanical microenvironment in directing cell fate and behavior.HighlightsGeneration of embryonic scale 3D morphogenesis using hydrogel encapsulationManipulating adhesion triggers emergence of specific morphologies and cell fatesAcquisition of germ layer cell fates mimics early human embryonic diversity
- Published
- 2020
- Full Text
- View/download PDF
32. Deep neural net tracking of human pluripotent stem cells reveals intrinsic behaviors directing morphogenesis
- Author
-
Ashley R.G. Libby, Todd C. McDevitt, and David A Joy
- Subjects
0301 basic medicine ,cell migration ,Cell ,Induced Pluripotent Stem Cells ,Morphogenesis ,neural net ,morphogenesis ,Cell Count ,Smad Proteins ,time-lapse imaging ,Bone Morphogenetic Protein 4 ,Cell fate determination ,Biology ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Genetics ,medicine ,Image Processing, Computer-Assisted ,Humans ,Cell Lineage ,human pluripotent stem cells ,Induced pluripotent stem cell ,cell segmentation ,Cells, Cultured ,Artificial neural network ,deep learning ,Cell migration ,Cell Differentiation ,Cell Biology ,differentiation ,Multicellular organism ,030104 developmental biology ,medicine.anatomical_structure ,cell tracking ,Neural Networks, Computer ,Neuroscience ,Developmental biology ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Summary Lineage tracing is a powerful tool in developmental biology to interrogate the evolution of tissue formation, but the dense, three-dimensional nature of tissue limits the assembly of individual cell trajectories into complete reconstructions of development. Human induced pluripotent stem cells (hiPSCs) can recapitulate aspects of developmental processes, providing an in vitro platform to assess the dynamic collective behaviors directing tissue morphogenesis. Here, we trained an ensemble of neural networks to track individual hiPSCs in time-lapse microscopy, generating longitudinal measures of cell and cellular neighborhood properties on timescales from minutes to days. Our analysis reveals that, while individual cell parameters are not strongly affected by pluripotency maintenance conditions or morphogenic cues, regional changes in cell behavior predict cell fate and colony organization. By generating complete multicellular reconstructions of hiPSC behavior, our tracking pipeline enables fine-grained understanding of morphogenesis by elucidating the role of regional behavior in early tissue formation., Highlights • An ensemble of neural nets segments hiPSC colonies with superhuman performanc • Heterotypic hiPSC phenotypes migrate divergently between colony center and edge • Colony size, substrate, and media alter hiPSC colony structure but not pluripotency • Morphogens induce distinct behaviors in hiPSCs despite convergent lineage patterning, In this article, Joy and colleagues report a cell segmentation and tracking pipeline employing an ensemble of convolutional neural networks to perform dense analysis of time-lapse imaging of human induced pluripotent stem cell colonies. They find distinctive behavioral signatures reflecting cell position, environmental cues, and morphogen treatment, enabling non-destructive monitoring of stem cell growth and differentiation.
- Published
- 2020
33. Deep neural net tracking of human pluripotent stem cells reveals intrinsic behaviors directing morphogenesis
- Author
-
David A Joy, Ashley R.G. Libby, and Todd C. McDevitt
- Subjects
Multicellular organism ,Artificial neural network ,Lineage tracing ,Morphogenesis ,Biology ,Induced pluripotent stem cell ,Neuroscience ,Convolutional neural network ,Developmental biology ,Time-lapse microscopy - Abstract
Lineage tracing is a powerful tool traditionally used in developmental biology to interrogate the evolutionary time course of tissue formation, but the dense, three-dimensional nature of tissue limits the ability to assemble individual traces into complete reconstructions of development. Human induced pluripotent stem cells (hiPSCs) enable recapitulation of various aspects of developmental processes, thereby providing an in vitro platform to assess the dynamic collective behaviors directing tissue morphogenesis. Here, we trained an ensemble of independent convolutional neural networks to identify individual hiPSCs imaged via time lapse microscopy in order to generate longitudinal measures of individual cell and dense cellular neighborhood properties simultaneously on timescales ranging from minutes to days. Our analysis reveals that while individual cell parameters are not strongly affected by extracellular microenvironmental conditions such as pluripotency maintenance regime or soluble morphogenic cues, regionally specific cell behaviors change in a manner predictive of organization dynamics. By generating complete multicellular reconstructions of hiPSC behavior, our cell tracking pipeline enables fine-grained understanding of developmental organization by elucidating the role of regional behavior stratification in early tissue formation.
- Published
- 2020
- Full Text
- View/download PDF
34. SARS-CoV-2 infection of human iPSC-derived cardiac cells predicts novel cytopathic features in hearts of COVID-19 patients
- Author
-
Will R. Flanigan, Todd C. McDevitt, Melanie Ott, Ken Nakamura, Gokul N. Ramadoss, Huihui Li, Jeffrey D. Whitman, David A Joy, Juan A. Perez-Bermejo, Camille R. Simoneau, Serah S Kang, Bruce R. Conklin, Ana Cannas da Silva, and Sarah J. Rockwood
- Subjects
Coronavirus ,Transcriptome ,Pathogenesis ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Biology ,Heart cells ,Fragmentation (cell biology) ,STM reports ,Virology ,Dna staining ,Reports ,Nuclear DNA - Abstract
Although coronavirus disease 2019 (COVID-19) causes cardiac dysfunction in up to 25% of patients, its pathogenesis remains unclear. Exposure of human induced pluripotent stem cell (iPSC)-derived heart cells to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) revealed productive infection and robust transcriptomic and morphological signatures of damage, particularly in cardiomyocytes. Transcriptomic disruption of structural genes corroborates adverse morphologic features, which included a distinct pattern of myofibrillar fragmentation and nuclear disruption. Human autopsy specimens from patients with COVID-19 reflected similar alterations, particularly sarcomeric fragmentation. These striking cytopathic features in cardiomyocytes provide insights into SARS-CoV-2-induced cardiac damage, offer a platform for discovery of potential therapeutics, and raise concerns about the long-term consequences of COVID-19 in asymptomatic as well as severe cases., Infection of human iPSC-derived cardiomyocytes by SARS-CoV-2 leads to specific cytopathic features reflected in patient autopsy samples.
- Published
- 2020
- Full Text
- View/download PDF
35. Cardiac Cell-Derived Matrices Impart Age-Specific Functional Properties to Human Cardiomyocytes
- Author
-
Kauss Ma, Whittaker Mn, Erica Stevenson, David A Joy, Nevan J. Krogan, Danielle L. Swaney, Ana Cannas da Silva, Sarah J. Rockwood, Todd C. McDevitt, and Mendoza-Camacho N
- Subjects
Extracellular matrix ,Surface coating ,Tissue culture ,Decellularization ,Collagen VI ,Chemistry ,Ficoll ,Adhesion ,Proteomics ,Cell biology - Abstract
Cell-derived matrices (CDMs) isolated from cultured cells provide complex and tissue-specific biochemical and physical cues derived from the extracellular matrix (ECM) that are lacking in typical tissue culture environments. However, current methods enhance ECM adhesion and thickness via introduction and promotion of singular matrix proteins, skewing the matrix composition, and confounding comparisons between CDMs. Here we developed a protocol that enhances CDM stability and deposition, respectively, by combining an L-polydopamine surface coating with Ficoll macromolecular crowing prior to hypotonic decellularization. This methodology was applied to the study of age-dependent phenotypic and functional changes observed in cardiac ECM by comparing the morphologic, electrophysiological and metabolic response of cardiomyocytes in response to CDMs produced by fetal and adult cardiac fibroblasts. Furthermore, mass spectrometry proteomics identified the enrichment of collagen VI in fetal CDMs, which we determined via siRNA-mediated silencing during CDM production to be necessary for maximal oxidative respiration in cardiomyocytes.
- Published
- 2020
- Full Text
- View/download PDF
36. Bi-directional Impacts of Heterotypic Interactions in Engineered 3D Human Cardiac Microtissues Revealed by Single-Cell RNA-Sequencing and Functional Analysis
- Author
-
David A Joy, Tracy A. Hookway, Oriane B. Matthys, Todd C. McDevitt, Reuben Thomas, and Jessica E. Sepulveda
- Subjects
Transcriptome ,Cell type ,Calcium imaging ,medicine.anatomical_structure ,Cell ,medicine ,Biology ,Cytoskeleton ,Gene ,Phenotype ,Function (biology) ,Cell biology - Abstract
Technological advancements have enabled the design of increasingly complex engineered tissue constructs, which better mimic native tissue cellularity. Therefore, dissecting the bi-directional interactions between distinct cell types in 3D is necessary to understand how heterotypic interactions at the single-cell level impact tissue-level properties. We systematically interrogated the interactions between cardiomyocytes (CMs) and cardiac non-myocytes in 3D self-assembled tissue constructs in an effort to determine the phenotypic and functional contributions of cardiac fibroblasts (CFs) and endothelial cells (ECs) to cardiac tissue properties. One week after tissue formation, cardiac microtissues containing CFs exhibited improved calcium handling function compared to microtissues comprised of CMs alone or CMs mixed with ECs, and CMs cultured with CFs exhibited distinct transcriptional profiles, with increased expression of cytoskeletal and ECM-associated genes. However, one month after tissue formation, functional and phenotypic differences between heterotypic tissues were mitigated, indicating diminishing impacts of non-myocytes on CM phenotype and function over time. The combination of single-cell RNA-sequencing and calcium imaging enabled the determination of reciprocal transcriptomic changes accompanying tissue-level functional properties in engineered heterotypic cardiac microtissues.
- Published
- 2020
- Full Text
- View/download PDF
37. Co-emergence of cardiac and gut tissues promotes cardiomyocyte maturation within human iPSC-derived organoids
- Author
-
Michael H. Lai, Andrew P. Blair, Oriane B. Matthys, David A Joy, Benoit G. Bruneau, Michael Alexanian, Ana C. Silva, Mara A. Kauss, Todd C. McDevitt, Vaishaali Natarajan, and Diwaker Turaga
- Subjects
Pluripotent Stem Cells ,Embryogenesis ,Endoderm ,Induced Pluripotent Stem Cells ,Morphogenesis ,Cell Differentiation ,Cell Biology ,Cell fate determination ,Biology ,Cell biology ,Organoids ,Paracrine signalling ,medicine.anatomical_structure ,Genetics ,medicine ,Organoid ,Molecular Medicine ,Humans ,Myocytes, Cardiac ,NODAL ,Induced pluripotent stem cell - Abstract
Summary During embryogenesis, paracrine signaling between tissues in close proximity contributes to the determination of their respective cell fate(s) and development into functional organs. Organoids are in vitro models that mimic organ formation and cellular heterogeneity, but lack the paracrine input of surrounding tissues. Here, we describe a human multilineage iPSC-derived organoid that recapitulates cooperative cardiac and gut development and maturation, with extensive cellular and structural complexity in both tissues. We demonstrate that the presence of endoderm tissue (gut/intestine) in the organoids contributed to the development of cardiac tissue features characteristic of stages after heart tube formation, including cardiomyocyte expansion, compartmentalization, enrichment of atrial/nodal cells, myocardial compaction, and fetal-like functional maturation. Overall, this study demonstrates the ability to generate and mature cooperative tissues originating from different germ lineages within a single organoid model, an advance that will further the examination of multi-tissue interactions during development, physiological maturation, and disease.
- Published
- 2020
38. Developmental co-emergence of cardiac and gut tissues modeled by human iPSC-derived organoids
- Author
-
Benoit G. Bruneau, Vaishaali Natarajan, Ana C. Silva, Michael Alexanian, Michael H. Lai, David A Joy, Diwakar Turaga, M.A. Kauss, Todd C. McDevitt, and Oriane B. Matthys
- Subjects
Paracrine signalling ,medicine.anatomical_structure ,Embryogenesis ,Organoid ,medicine ,Tissue formation ,Endoderm ,Compartmentalization (psychology) ,Biology ,NODAL ,In vitro ,Cell biology - Abstract
During embryogenesis, paracrine signaling between tissues in close proximity contributes to the determination of their respective cell fate(s) and development into functional organs. Organoids arein vitromodels that mimic organ formation and cellular heterogeneity, but lack the paracrine input of surrounding tissues. Here, we describe a human multilineage iPSC-derived organoid that recapitulates cooperative cardiac and gut development and displays extensive cellular and structural complexity of both tissues. We demonstrate that the presence of endoderm tissue (gut/intestine) in multilineage organoids contributed to the development of the cardiac tissue, specifically cardiomyocyte expansion, compartmentalization, enrichment of atrial/nodal cells, myocardial compaction and functional fetal-like maturation. Overall, this study demonstrates the ability to generate specific cooperative tissues originating from different germ lineages within a single organoid model, an advance that will further the examination of multi-tissue interactions during development and disease.
- Published
- 2020
- Full Text
- View/download PDF
39. Axial Elongation of Caudalized Human Organoids Mimics Neural Tube Development
- Author
-
NH Elder, Martina Z Krakora, EA Gaylord, Jessica C Butts, David A Joy, Arg Libby, Todd C. McDevitt, Frederico N. Mendoza-Camacho, and Emily A. Bulger
- Subjects
Neuroepithelial cell ,medicine.anatomical_structure ,SOX2 ,embryonic structures ,Embryogenesis ,Neural tube ,medicine ,Paraxial mesoderm ,Organoid ,Wnt signaling pathway ,Biology ,Noggin ,Cell biology - Abstract
Axial elongation of the neural tube is critical during mammalian embryogenesis to establish the anterior-posterior body axis1, but this process is difficult to interrogate directly because it occurs post-implantation2,3. Here we report an organoid model of neural tube extension by caudalized human pluripotent stem cell (hPSC) aggregates that recapitulates the morphologic and temporal gene expression patterns of neural tube development. Axially extending organoids consisting largely of longitudinally elongated neuroepithelial compartments also contained TBXT(+)SOX2(+) neuromesodermal progenitors, PAX6(+)nestin(+) neural progenitor populations, and MEOX1(+) paraxial mesoderm populations. Wnt agonism stimulated singular axial extensions in a dose-dependent manner, and elongated organoids displayed regionalized rostral-caudal HOX gene expression, with spatially distinct hindbrain (HOXB1) expression from brachial (HOXC6) and thoracic (HOXB9) regions. CRISPR-interference-mediated silencing of the TBXT, a downstream Wnt target, increased neuroepithelial compartmentalization and resulted in multiple extensions per aggregate. Further, knock-down of BMP inhibitors, Noggin and Chordin, induced elongation phenotypes that mimicked murine knockout models. These results indicate the potent morphogenic capacity of caudalized hPSC organoids to undergo axial elongation in a manner that can be used to dissect the cellular organization and patterning decisions that dictate early nervous system development in humans.
- Published
- 2020
- Full Text
- View/download PDF
40. Single-Cell Determination of Cardiac Microtissue Structure and Function Using Light Sheet Microscopy
- Author
-
David A Joy, Meredith E. K. Calvert, Diwakar Turaga, Todd C. McDevitt, Tracy A. Hookway, and Oriane B. Matthys
- Subjects
education.field_of_study ,Cell type ,Tissue Engineering ,Chemistry ,Population ,Cell ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Cell Communication ,Fibroblasts ,Structure and function ,Multicellular organism ,Calcium imaging ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Live cell imaging ,Light sheet fluorescence microscopy ,Biophysics ,medicine ,Humans ,Calcium ,Myocytes, Cardiac ,Single-Cell Analysis ,education - Abstract
Native cardiac tissue is composed of heterogeneous cell populations that work cooperatively for proper tissue function; thus, engineered tissue models have moved toward incorporating multiple cardiac cell types in an effort to recapitulate native multicellular composition and organization. Cardiac tissue models composed of stem cell-derived cardiomyocytes (CMs) require inclusion of non-myocytes to promote stable tissue formation, yet the specific contributions of the supporting non-myocyte population on the parenchymal CMs and cardiac microtissues have to be fully dissected. This gap can be partly attributed to limitations in technologies able to accurately study the individual cellular structure and function that comprise intact three-dimensional (3D) tissues. The ability to interrogate the cell-cell interactions in 3D tissue constructs has been restricted by conventional optical imaging techniques that fail to adequately penetrate multicellular microtissues with sufficient spatial resolution. Light sheet fluorescence microscopy (LSFM) overcomes these constraints to enable single-cell resolution structural and functional imaging of intact cardiac microtissues. Multicellular spatial distribution analysis of heterotypic cardiac cell populations revealed that CMs and cardiac fibroblasts were randomly distributed throughout 3D microtissues. Furthermore, calcium imaging of live cardiac microtissues enabled single-cell detection of CM calcium activity, which showed that functional heterogeneity correlated with spatial location within the tissues. This study demonstrates that LSFM can be utilized to determine single-cell spatial and functional interactions of multiple cell types within intact 3D engineered microtissues, thereby facilitating the determination of structure-function relationships at both tissue-level and single-cell resolution. Impact statement The ability to achieve single-cell resolution by advanced three-dimensional light imaging techniques enables exquisite new investigation of multicellular analyses in native and engineered tissues. In this study, light sheet fluorescence microscopy was used to define structure-function relationships of distinct cell types in engineered cardiac microtissues by determining heterotypic cell distributions and interactions throughout the tissues as well as by assessing regional differences in calcium handing functional properties at the individual cardiomyocyte level.
- Published
- 2020
41. Bioengineered optogenetic model of human neuromuscular junction
- Author
-
Jennifer M. Colón-Mercado, Michael E. Ward, Todd C. McDevitt, Carissa M. Feliciano, Yihuai Qu, Bruce R. Conklin, Gordana Vunjak-Novakovic, Matthew Carter, Stephen P. Ma, Trevor R. Nash, Lyandysha V. Zholudeva, Miguel Chavez, Keith Yeager, Roger D. Kamm, Luke M. Judge, Olaia F. Vila, and Carmen Lai
- Subjects
Induced Pluripotent Stem Cells ,Neuromuscular Junction ,Biophysics ,Bioengineering ,Optogenetics ,Article ,Neuromuscular junction ,Biomaterials ,Blood serum ,Tissue engineering ,medicine ,Humans ,Muscle, Skeletal ,Induced pluripotent stem cell ,business.industry ,Reproducibility of Results ,Skeletal muscle ,Small sample ,medicine.disease ,Myasthenia gravis ,medicine.anatomical_structure ,Mechanics of Materials ,Ceramics and Composites ,business ,Neuroscience - Abstract
Functional human tissues engineered from patient-specific induced pluripotent stem cells (hiPSCs) hold great promise for investigating the progression, mechanisms, and treatment of musculoskeletal diseases in a controlled and systematic manner. For example, bioengineered models of innervated human skeletal muscle could be used to identify novel therapeutic targets and treatments for patients with complex central and peripheral nervous system disorders. There is a need to develop standardized and objective quantitative methods for engineering and using these complex tissues, in order increase their robustness, reproducibility, and predictiveness across users. Here we describe a standardized method for engineering an isogenic, patient specific human neuromuscular junction (NMJ) that allows for automated quantification of NMJ function to diagnose disease using a small sample of blood serum and evaluate new therapeutic modalities. By combining tissue engineering, optogenetics, microfabrication, optoelectronics and video processing, we created a novel platform for the precise investigation of the development and degeneration of human NMJ. We demonstrate the utility of this platform for the detection and diagnosis of myasthenia gravis, an antibody-mediated autoimmune disease that disrupts the NMJ function.
- Published
- 2021
- Full Text
- View/download PDF
42. Mouse gastruloids take heart
- Author
-
Deepak Srivastava and Todd C. McDevitt
- Subjects
0301 basic medicine ,business.industry ,Cell ,Organogenesis ,030204 cardiovascular system & hematology ,Embryonic stem cell ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Organoid ,Medicine ,Cardiology and Cardiovascular Medicine ,business ,Ex vivo - Abstract
Mouse embryonic organoids that model cardiac development ex vivo could be used as a high-throughput, experimentally tractable system to evaluate crucial cell populations and environmental factors that contribute to normal and abnormal cardiogenesis.
- Published
- 2021
- Full Text
- View/download PDF
43. Single cell determination of cardiac microtissue structure and function using light sheet microscopy
- Author
-
Todd C. McDevitt, David A Joy, Meredith E. K. Calvert, Diwakar Turaga, Tracy A. Hookway, and Oriane B. Matthys
- Subjects
Cell type ,education.field_of_study ,Chemistry ,Population ,Cell ,Structure and function ,Cell biology ,Multicellular organism ,Cell determination ,Calcium imaging ,medicine.anatomical_structure ,Light sheet fluorescence microscopy ,medicine ,education - Abstract
Native cardiac tissue is comprised of heterogeneous cell populations that work cooperatively for proper tissue function; thus, engineered tissue models have moved toward incorporating multiple cardiac cell types in an effort to recapitulate native multicellular composition and organization. Cardiac tissue models comprised of stem cell-derived cardiomyocytes require inclusion of non-myocytes to promote stable tissue formation, yet the specific contributions of the supporting non-myocyte population on the parenchymal cardiomyocytes and cardiac microtissues have yet to be fully dissected. This gap can be partly attributed to limitations in technologies able to accurately study the individual cellular structure and function that comprise intact 3D tissues. The ability to interrogate the cell-cell interactions in 3D tissue constructs has been restricted by conventional optical imaging techniques that fail to adequately penetrate multicellular microtissues with sufficient spatial resolution. Light sheet fluorescence microscopy overcomes these constraints to enable single cell-resolution structural and functional imaging of intact cardiac microtissues. Multicellular spatial distribution analysis of heterotypic cardiac cell populations revealed that cardiomyocytes and cardiac fibroblasts were randomly distributed throughout 3D microtissues. Furthermore, calcium imaging of live cardiac microtissues enabled single-cell detection of cardiomyocyte calcium activity, which showed that functional heterogeneity correlated with spatial location within the tissues. This study demonstrates that light sheet fluorescence microscopy can be utilized to determine single-cell spatial and functional interactions of multiple cell types within intact 3D engineered microtissues, thereby facilitating the determination of structure-function relationships at both tissue-level and single-cell resolution.Impact StatementThe ability to achieve single-cell resolution by advanced 3D light imaging techniques enables exquisite new investigation of multicellular analyses in native and engineered tissues. In this study, light sheet fluorescence microscopy was used to define structure-function relationships of distinct cell types in engineered cardiac microtissues by determining heterotypic cell distributions and interactions throughout the tissues as well as by assessing regional differences in calcium handing functional properties at the individual cardiomyocyte level.
- Published
- 2020
- Full Text
- View/download PDF
44. Heparin-mediated delivery of bone morphogenetic protein-2 improves spatial localization of bone regeneration
- Author
-
Laxminarayanan Krishnan, Tel Rouse, Catherine Chou, Marian H. Hettiaratchi, Todd C. McDevitt, and Robert E. Guldberg
- Subjects
Scaffold ,Bone Regeneration ,animal structures ,medicine.medical_treatment ,Bone Morphogenetic Protein 2 ,02 engineering and technology ,Ossification ,Bone morphogenetic protein 2 ,03 medical and health sciences ,Drug Delivery Systems ,In vivo ,Transforming Growth Factor beta ,Osteogenesis ,medicine ,Animals ,Humans ,Spatial localization ,Health and Medicine ,Femur ,Bone regeneration ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,5.2 Cellular and gene therapies ,Chemistry ,Heparin ,Ossification, Heterotopic ,SciAdv r-articles ,X-Ray Microtomography ,021001 nanoscience & nanotechnology ,medicine.disease ,Stem Cell Research ,Recombinant Proteins ,Cell biology ,Rats ,Applied Sciences and Engineering ,Spinal fusion ,Musculoskeletal ,embryonic structures ,Heterotopic ossification ,Heterotopic ,Stem Cell Research - Nonembryonic - Non-Human ,Collagen ,Development of treatments and therapeutic interventions ,0210 nano-technology ,Research Article ,medicine.drug - Abstract
Heparin-based microparticles improve spatial localization of BMP-2 delivery, decreasing bone formation outside of the injury site., Supraphysiologic doses of bone morphogenetic protein-2 (BMP-2) are used clinically to promote bone formation in fracture nonunions, large bone defects, and spinal fusion. However, abnormal bone formation (i.e., heterotopic ossification) caused by rapid BMP-2 release from conventional collagen sponge scaffolds is a serious complication. We leveraged the strong affinity interactions between heparin microparticles (HMPs) and BMP-2 to improve protein delivery to bone defects. We first developed a computational model to investigate BMP-2–HMP interactions and demonstrated improved in vivo BMP-2 retention using HMPs. We then evaluated BMP-2–loaded HMPs as a treatment strategy for healing critically sized femoral defects in a rat model that displays heterotopic ossification with clinical BMP-2 doses (0.12 mg/kg body weight). HMPs increased BMP-2 retention in vivo, improving spatial localization of bone formation in large bone defects and reducing heterotopic ossification. Thus, HMPs provide a promising opportunity to improve the safety profile of scaffold-based BMP-2 delivery.
- Published
- 2020
45. Imaging the developing human external and internal urogenital organs with light sheet fluorescence microscopy
- Author
-
Mei Cao, Todd C. McDevitt, Meredith E. K. Calvert, Gerald R. Cunha, Laurence S. Baskin, Dylan Isaacson, Adriane Sinclair, Yi Li, Joel Shen, and Dylan A. McCreedy
- Subjects
0301 basic medicine ,Internal genitalia ,Urologic Diseases ,Cancer Research ,Microscope ,Image Processing ,Urogenital development ,High resolution ,Urogenital System ,Biology ,Fluorescence ,law.invention ,Imaging ,Specimen Handling ,Paediatrics and Reproductive Medicine ,03 medical and health sciences ,Imaging, Three-Dimensional ,0302 clinical medicine ,Computer-Assisted ,law ,Microscopy ,Image Processing, Computer-Assisted ,Humans ,Molecular Biology ,Pediatric ,Light sheet fluorescence microscopy ,Cell Biology ,030104 developmental biology ,Selective plane illumination microscopy ,Microscopy, Fluorescence ,Genital development ,External genitalia ,CLARITY ,Human fetal ,Three-Dimensional ,Biochemistry and Cell Biology ,030217 neurology & neurosurgery ,Clearance ,Biomedical engineering ,Developmental Biology - Abstract
Technological advances in three-dimensional (3D) reconstruction techniques have previously enabled paradigm shifts in our understanding of human embryonic and fetal development. Light sheet fluorescence microscopy (LSFM) is a recently-developed technique that uses thin planes of light to optically section whole-mount cleared and immunolabeled biologic specimens. The advent of commercially-available light sheet microscopes has facilitated a new generation of research into protein localization and tissue dynamics at extremely high resolution. Our group has applied LSFM to study developing human fetal external genitalia, internal genitalia and kidneys. This review describes LSFM and presents our group's technique for preparing, clearing, immunostaining and imaging human fetal urogenital specimens. We then present light sheet images and videos of each element of the developing human urogenital system. To the extent of our knowledge, the work conducted by our laboratory represents the first description of a method for performing LSFM on the full human urogenital system during the embryonic and fetal periods.
- Published
- 2020
46. V2a Interneuron Differentiation from Mouse and Human Pluripotent Stem Cells
- Author
-
Nisha Iyer, Russell E. Thompson, Nick White, Shelly E. Sakiyama-Elbert, Jessica C. Butts, and Todd C. McDevitt
- Subjects
Pluripotent Stem Cells ,Interneuron ,Bioinformatics ,Neurogenesis ,Cellular differentiation ,Cell Culture Techniques ,Hindbrain ,Biology ,Regenerative Medicine ,Medical and Health Sciences ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Interneurons ,Stem Cell Research - Nonembryonic - Human ,medicine ,Animals ,Humans ,Stem Cell Research - Embryonic - Human ,Induced pluripotent stem cell ,030304 developmental biology ,0303 health sciences ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,Biological Sciences ,Stem Cell Research ,Spinal cord ,Phenotype ,medicine.anatomical_structure ,nervous system ,Cell culture ,Chemical Sciences ,Generic health relevance ,Neuroscience ,030217 neurology & neurosurgery - Abstract
V2a interneurons are located in the hindbrain and spinal cord, where they provide rhythmic input to major motor control centers. Many of the phenotypic properties and functions of excitatory V2a interneurons have yet to be fully defined. Definition of these properties could lead to novel regenerative therapies for traumatic injuries and drug targets for chronic degenerative diseases. Here we describe how to produce V2a interneurons from mouse and human pluripotent stem cells (PSCs), as well as strategies to characterize and mature the cells for further analysis. The described protocols are based on a sequence of small-molecule treatments that induce differentiation of PSCs into V2a interneurons. We also include a detailed description of how to phenotypically characterize, mature, and freeze the cells. The mouse and human protocols are similar in regard to the sequence of small molecules used but differ slightly in the concentrations and durations necessary for induction. With the protocols described, scientists can expect to obtain V2a interneurons with purities of ~75% (mouse) in 7 d and ~50% (human) in 20 d. V2a interneurons are differentiated from mouse and human pluripotent stem cells following culture in the presence of a sequence of small-molecule treatments.
- Published
- 2019
47. Hyaluronic acid as a macromolecular crowding agent for production of cell-derived matrices
- Author
-
Katja Schenke-Layland, A.J. Rickards, Tanja Dominko, M.A. Kauss, Marsha W. Rolle, W. Linthicum, Dalia Shendi, Q. Wen, Julia Marzi, Todd C. McDevitt, David Dolivo, and Silke Keller
- Subjects
Indoles ,Macromolecular Substances ,Polymers ,0206 medical engineering ,Biomedical Engineering ,Ficoll ,02 engineering and technology ,Spectrum Analysis, Raman ,Biochemistry ,Biomaterials ,Extracellular matrix ,chemistry.chemical_compound ,Hyaluronic acid ,medicine ,Animals ,Humans ,Hyaluronic Acid ,Fibroblast ,Molecular Biology ,Cells, Cultured ,Chemistry ,Viscosity ,Infant, Newborn ,Biomaterial ,General Medicine ,Fibroblasts ,021001 nanoscience & nanotechnology ,020601 biomedical engineering ,Matrix Metalloproteinases ,Extracellular Matrix ,Fibronectins ,medicine.anatomical_structure ,Gene Expression Regulation ,Solubility ,Cell culture ,Biophysics ,Cattle ,Collagen ,Laminin ,0210 nano-technology ,Wound healing ,Macromolecular crowding ,Biotechnology - Abstract
Cell-derived matrices (CDMs) provide an exogenous source of human extracellular matrix (ECM), with applications as cell delivery vehicles, substrate coatings for cell attachment and differentiation, and as biomaterial scaffolds. However, commercial application of CDMs has been hindered due to the prolonged culture time required for sufficient ECM accumulation. One approach to increasing matrix deposition in vitro is macromolecular crowding (MMC), which is a biophysical phenomenon that limits the diffusion of ECM precursor proteins, resulting in increased ECM accumulation at the cell layer. Hyaluronic acid (HA), a natural MMC highly expressed in vivo during fetal development, has been shown to play a role in ECM production, but has not been investigated as a macromolecule for increasing cell-mediated ECM deposition in vitro. In the current study, we hypothesized that HA can act as a MMC, and increase cell-mediated ECM production. Human dermal fibroblasts were cultured for 3, 7, or 14 days with 0%, 0.05%, or 0.5% high molecular weight HA. Ficoll 70/400 was used as a positive control. SDS-PAGE, Sircol, and hydroxyproline assays indicated that 0.05% HA-treated cultures had significantly higher mean collagen deposition at 14 days, whereas Ficoll 70/400-treated cultures had significantly lower collagen production compared to the HA and untreated controls. However, fluorescent immunostaining of ECM proteins and quantification of mean gray values did not indicate statistically significant differences in ECM production in HA or Ficoll 70/400-treated cultures compared to untreated controls. Raman imaging (a marker-free spectral imaging method) indicated that HA increased ECM deposition in human dermal fibroblasts. These results are consistent with decreases in CDM stiffness observed in Ficoll 70/400-treated cultures by atomic force microscopy. Overall, these results indicate that there are macromolecule- and cell type- dependent effects on matrix assembly, turnover, and stiffness in cell-derived matrices. STATEMENT OF SIGNIFICANCE: Cell-derived matrices (CDMs) are versatile biomaterials with many regenerative medicine applications, including as cell and drug delivery vehicles and scaffolds for wound healing and tissue regeneration. While CDMs have several advantages, their commercialization has been limited due to the prolonged culture time required to achieve CDM synthesis in vitro. In this study, we explored the use of hyaluronic acid (HA) as a macromolecular crowder in human fibroblast cell cultures to support production of CDM biomaterials. Successful application of macromolecular crowding will allow development of human cell-derived, xeno-free biomaterials that re-capitulate the native human tissue microenvironment.
- Published
- 2019
48. Phenotypic Variation Between Stromal Cells Differentially Impacts Engineered Cardiac Tissue Function
- Author
-
David A Joy, Jessica E. Sepulveda, Tracy A. Hookway, Oriane B. Matthys, Sarah Rains, Federico N Mendoza-Camacho, and Todd C. McDevitt
- Subjects
Male ,Special Issue Articles ,Cell type ,Stromal cell ,0206 medical engineering ,Biomedical Engineering ,cardiomyocytes ,Bioengineering ,02 engineering and technology ,Biology ,Cardiovascular ,Regenerative Medicine ,Biochemistry ,Biomaterials ,03 medical and health sciences ,heterotypic interactions ,Tissue engineering ,cardiac microtissues ,2.1 Biological and endogenous factors ,Myocyte ,Humans ,Myocytes, Cardiac ,Aetiology ,030304 developmental biology ,Myocytes ,0303 health sciences ,stem cell-derived tissues ,Stem Cell Research - Induced Pluripotent Stem Cell ,Heart development ,Tissue Engineering ,Myocardium ,Materials Engineering ,Fibroblasts ,Stem Cell Research ,020601 biomedical engineering ,Phenotype ,In vitro ,Cell biology ,Heart Disease ,Female ,Biochemistry and Cell Biology ,Stromal Cells ,Cardiac ,Function (biology) - Abstract
Throughout heart development, cardiomyocytes differentiate and mature in direct contact with nonparenchymal cell types, such as cardiac fibroblasts. Thus, when modeling myocardial tissue in vitro, tissue engineers include a supporting stromal cell population that is necessary for tissue formation, although the source of stromal cells has varied widely. This study systematically characterized the phenotype of commonly used stromal cell populations and analyzed the differential impacts of stromal phenotype on cardiac microtissue phenotype and function. Quantitative morphometric analysis, flow cytometry, unbiased morphological feature clustering, and RNA sequencing of the different stromal populations revealed variable cell morphologies, surface marker expression, and gene signatures, with primary adult stromal populations exhibiting more similar phenotypes to each other than to stem cell-derived and progenitor populations. The ability of self-assembled cardiac microtissues to consistently form tissues was highly dependent on the stromal population mixed with stem cell-derived cardiomyocytes, with cardiac fibroblasts and dermal fibroblasts (DFs) forming the most robust tissues compared with mesenchymal stromal cells and induced pluripotent stem cell-derived fibroblasts. Cardiac fibroblasts and DFs also resulted in cardiac microtissues displaying a more mature calcium handling profile, with increased amplitude and upstroke velocity. These results demonstrate the breadth of phenotypic variation across stromal populations owing to cell and tissue source, with certain primary populations, such as cardiac fibroblasts and DFs, supporting cardiac microtissue phenotype and improved calcium handling function. IMPACT STATEMENT: Understanding the relationship between parenchymal and supporting cell populations is paramount to recapitulate the multicellular complexity of native tissues. Incorporation of stromal cells is widely recognized to be necessary for the stable formation of stem cell-derived cardiac tissues; yet, the types of stromal cells used have varied widely. This study systematically characterized several stromal populations and found that stromal phenotype and morphology was highly variable depending on cell source and exerted differential impacts on cardiac tissue function and induced pluripotent stem cell–cardiomyocyte phenotype. Therefore, the choice of supporting stromal population can differentially impact the phenotypic or functional performance of engineered cardiac tissues.
- Published
- 2019
49. Comparable Decellularization of Fetal and Adult Cardiac Tissue Explants as 3D-like Platforms for In Vitro Studies
- Author
-
Ana C. Silva, Maria José Oliveira, Perpétua Pinto-do-Ó, Diana S. Nascimento, Todd C. McDevitt, and Mário A. Barbosa
- Subjects
0301 basic medicine ,Adult ,General Chemical Engineering ,Cell Culture Techniques ,Fetal heart ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Extracellular matrix ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Fetus ,Tissue engineering ,Animals ,Humans ,Decellularization ,General Immunology and Microbiology ,Tissue Engineering ,General Neuroscience ,Myocardium ,Tissue explants ,In vitro ,Cell biology ,Extracellular Matrix ,Surface coating ,030104 developmental biology ,030220 oncology & carcinogenesis - Abstract
Current knowledge of extracellular matrix (ECM)-cell communication translates to large two-dimensional (2D) in vitro culture studies where ECM components are presented as a surface coating. These culture systems constitute a simplification of the complex nature of the tissue ECM that encompasses biochemical composition, structure, and mechanical properties. To better emulate the ECM-cell communication shaping the cardiac microenvironment, we developed a protocol that allows for the decellularization of the whole fetal heart and adult left ventricle tissue explants simultaneously for comparative studies. The protocol combines the use of a hypotonic buffer, a detergent of anionic surfactant properties, and DNase treatment without any requirement for specialized skills or equipment. The application of the same decellularization strategy across tissue samples from subjects of various age is an alternative approach to perform comparative studies. The present protocol allows the identification of unique structural differences across fetal and adult cardiac ECM mesh and biological cellular responses. Furthermore, the herein methodology demonstrates a broader application being successfully applied in other tissues and species with minor adjustments, such as in human intestine biopsies and mouse lung.
- Published
- 2019
50. A microfluidic trap array for longitudinal monitoring and multi-modal phenotypic analysis of individual stem cell aggregates
- Author
-
Emily Jackson-Holmes, Hang Lu, and Todd C. McDevitt
- Subjects
0301 basic medicine ,Microfluidics ,Biomedical Engineering ,Morphogenesis ,Fluorescent Antibody Technique ,Bioengineering ,Cell Separation ,Computational biology ,Biology ,Biochemistry ,Article ,Trap (computing) ,Mice ,03 medical and health sciences ,Phenotypic analysis ,Animals ,Induced pluripotent stem cell ,Embryonic Stem Cells ,Cell Aggregation ,Disease mechanisms ,Equipment Design ,General Chemistry ,Multiple modes ,Microfluidic Analytical Techniques ,Phenotype ,030104 developmental biology ,Stem cell ,Biomedical engineering - Abstract
Three-dimensional pluripotent stem cell (PSC) cultures have the ability to undergo differentiation, self-organization, and morphogenesis to yield complex, in vitro tissue models that recapitulate key elements of native tissues. These tissue models offer a system for studying mechanisms of tissue development, investigating disease mechanisms, and performing drug screening. It remains challenging, however, to standardize PSC aggregate differentiation and morphogenesis methods due to heterogeneity stemming from biological and environmental sources. It is also difficult to monitor and assess large numbers of individual samples longitudinally throughout culture using typical batch-based culture methods. To address these challenges, we have developed a microfluidic platform for culture, longitudinal monitoring, and phenotypic analysis of individual stem cell aggregates. This platform uses a hydrodynamic loading principle to capture pre-formed stem cell aggregates in independent traps. We demonstrated that multi-day culture of aggregates in this platform reduces heterogeneity in phenotypic parameters such as size and morphology. Additionally, we showed that culture and analysis steps can be performed sequentially in the same platform, enabling correlation of multiple modes of analysis for individual samples. We anticipate this platform being applied to improve abilities for phenotypic analysis of PSC aggregate tissues and to facilitate research in standardizing culture systems in order to dually increase the yield and reduce the heterogeneity of PSC-derived tissues.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.